Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters










Publication year range
1.
Poult Sci ; 83(12): 1973-8, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15615009

ABSTRACT

Immunization using a plasmid to deliver an encoded protein for expression in situ as the antigen is a promising technology. A plasmid encoding the enterotoxigenic Escherichia coli K88 fimbrial protein FaeG when injected into chickens stimulates the production of antibodies against the fimbrial protein, similar to what has been observed in mice. The efficacy of a genetic adjuvant on fimbrial antibody production was tested by introducing the gene for chicken interleukin-6 in tandem with the faeG gene. Expression of both the fimbrial FaeG protein and chicken interleukin-6 protein was confirmed in COS-M6 cells. Slightly higher antiFaeG antibody titer in chickens was obtained compared with immunization with the plasmid encoding FaeG alone, especially at 10 (19%, P < 0.05) and 12 (27%, P < 0.05) wk, respectively, after the secondary immunization. Elevated antiFaeG antibody titer induced by chicken interleukin-6 and FaeG proteins expressed jointly persisted longer than when induced by FaeG protein alone. This is the first report of an avian cytokine enhancing an immune response, and confirms that coexpression of the antigen and adjuvant from a plasmid delivered by DNA immunization is an effective protocol.


Subject(s)
Adhesins, Escherichia coli/genetics , Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/immunology , Chickens/immunology , Escherichia coli Proteins/immunology , Fimbriae Proteins/immunology , Interleukin-6/genetics , Plasmids/immunology , Adhesins, Escherichia coli/administration & dosage , Adhesins, Escherichia coli/immunology , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Chickens/genetics , DNA, Bacterial/immunology , Egg Yolk/immunology , Escherichia coli/immunology , Escherichia coli Proteins/administration & dosage , Escherichia coli Proteins/genetics , Female , Fimbriae Proteins/administration & dosage , Fimbriae Proteins/genetics , Gene Expression Regulation , Immunization/veterinary , Interleukin-6/administration & dosage , Interleukin-6/immunology , Ovum/immunology , Plasmids/administration & dosage , Plasmids/genetics , Time Factors
2.
Cell Mol Life Sci ; 61(2): 192-208, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14745498

ABSTRACT

More than 300 catalase sequences are now available, divided among monofunctional catalases (> 225), bifunctional catalase-peroxidases (> 50) and manganese-containing catalases (> 25). When combined with the recent appearance of crystal structures from at least two representatives from each of these groups (nine from the monofunctional catalases), valuable insights into the catalatic reaction mechanism in its various forms and into catalase evolution have been gained. The structures have revealed an unusually large number of modifications unique to catalases, a result of interacting with reactive oxygen species. Biochemical and physiological characterization of catalases from many different organisms has revealed a surprisingly wide range of catalatic efficiencies, despite similar sequences. Catalase gene expression in micro-organisms generally is controlled either by sensors of reactive oxygen species or by growth phase regulons, although the detailed mechanisms vary considerably.


Subject(s)
Catalase/chemistry , Catalase/physiology , Bacteria/enzymology , Enzyme Stability , Fungi/enzymology , Gene Expression Regulation/physiology , Heme/chemistry , Kinetics , Phylogeny , Protein Structure, Quaternary
3.
Proteins ; 44(3): 270-81, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11455600

ABSTRACT

The active site of heme catalases is buried deep inside a structurally highly conserved homotetramer. Channels leading to the active site have been identified as potential routes for substrate flow and product release, although evidence in support of this model is limited. To investigate further the role of protein structure and molecular channels in catalysis, the crystal structures of four active site variants of catalase HPII from Escherichia coli (His128Ala, His128Asn, Asn201Ala, and Asn201His) have been determined at approximately 2.0-A resolution. The solvent organization shows major rearrangements with respect to native HPII, not only in the vicinity of the replaced residues but also in the main molecular channel leading to the heme distal pocket. In the two inactive His128 variants, continuous chains of hydrogen bonded water molecules extend from the molecular surface to the heme distal pocket filling the main channel. The differences in continuity of solvent molecules between the native and variant structures illustrate how sensitive the solvent matrix is to subtle changes in structure. It is hypothesized that the slightly larger H(2)O(2) passing through the channel of the native enzyme will promote the formation of a continuous chain of solvent and peroxide. The structure of the His128Asn variant complexed with hydrogen peroxide has also been determined at 2.3-A resolution, revealing the existence of hydrogen peroxide binding sites both in the heme distal pocket and in the main channel. Unexpectedly, the largest changes in protein structure resulting from peroxide binding are clustered on the heme proximal side and mainly involve residues in only two subunits, leading to a departure from the 222-point group symmetry of the native enzyme. An active role for channels in the selective flow of substrates through the catalase molecule is proposed as an integral feature of the catalytic mechanism. The Asn201His variant of HPII was found to contain unoxidized heme b in combination with the proximal side His-Tyr bond suggesting that the mechanistic pathways of the two reactions can be uncoupled.


Subject(s)
Catalase/chemistry , Escherichia coli/enzymology , Hydrogen Peroxide/chemistry , Alleles , Asparagine/genetics , Binding Sites , Catalase/metabolism , Catalysis , Crystallization , Heme/metabolism , Histamine/genetics , Hydrogen Peroxide/metabolism , Models, Molecular , Mutation , Oxidation-Reduction , Solvents/chemistry
4.
Acta Crystallogr D Biol Crystallogr ; 57(Pt 8): 1184-6, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11468413

ABSTRACT

Haem-containing catalases are homotetrameric molecules that degrade hydrogen peroxide. Phylogenetically, the haem-containing catalases can be grouped into three main lines or clades. The crystal structures of seven catalases have been determined, all from clades II and III. In order to obtain a structure of an enzyme from clade I, which includes all plant, algae and some bacterial enzymes, two bacterial catalases, CatF from Pseudomonas syringae and Kat from Listeria seeligeri, have been crystallized by the hanging-drop vapour-diffusion technique, using PEG and ammonium sulfate as precipitants, respectively. Crystals of P. syringae CatF, with a plate-like morphology, belong to the monoclinic space group P2(1), with unit-cell parameters a = 60.6, b = 153.9, c = 109.2 A, beta = 102.8 degrees. From these crystals a diffraction data set to 1.8 A resolution with 98% completeness was collected using synchrotron radiation. Crystals of L. seeligeri Kat, with a well developed bipyramidal morphology, belong to space group I222 (or I2(1)2(1)2(1)), with unit-cell parameters a = 74.4, b = 121.3, c = 368.5 A. These crystals diffracted beyond 2.2 A resolution when using synchrotron radiation, but presented anisotropic diffraction, with the weakest direction perpendicular to the long c axis.


Subject(s)
Catalase/chemistry , Listeria/enzymology , Pseudomonas/enzymology , Crystallization , Crystallography, X-Ray , Protein Conformation
5.
Biochem Soc Trans ; 29(Pt 2): 99-105, 2001 May.
Article in English | MEDLINE | ID: mdl-11356135

ABSTRACT

Catalase-peroxidases are bifunctional peroxidases exhibiting an overwhelming catalase activity and a substantial peroxidase activity. Here we present a kinetic study of the formation and reduction of the key intermediate compound I by probing the role of the conserved tryptophan at the distal haem cavity site. Two wild-type proteins and three mutants of Synechocystis catalase-peroxidase (W122A and W122F) and Escherichia coli catalase-peroxidase (W105F) have been investigated by steady-state and stopped-flow spectroscopy. W122F and W122A completely lost their catalase activity whereas in W105F the catalase activity was reduced by a factor of about 5000. However, the mutations did not influence both formation of compound I and its reduction by peroxidase substrates. It was demonstrated unequivocally that the rate of compound I reduction by pyrogallol or o-dianisidine sometimes even exceeded that of the wild-type enzyme. This study demonstrates that the indole ring of distal Trp in catalase-peroxidases is essential for the two-electron reduction of compound I by hydrogen peroxide but not for compound I formation or for peroxidase reactivity (i.e. the one-electron reduction of compound I).


Subject(s)
Bacterial Proteins , Cyanobacteria/enzymology , Escherichia coli/enzymology , Peroxidases/metabolism , Tryptophan/metabolism , Binding Sites , Catalysis , Cyanobacteria/genetics , Cytochrome-c Peroxidase/chemistry , Cytochrome-c Peroxidase/metabolism , Escherichia coli/genetics , Heme/metabolism , Hydrogen Peroxide/metabolism , Kinetics , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutation/genetics , Oxidation-Reduction , Peroxidases/chemistry , Peroxidases/genetics , Protein Binding , Spectrophotometry , Tryptophan/genetics , Yeasts/enzymology
6.
Biochim Biophys Acta ; 1546(1): 44-54, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11257507

ABSTRACT

The catalase-peroxidase encoded by katG of Mycobacterium tuberculosis is a more effective activator of the antibiotic isoniazid than is the equivalent enzyme from Escherichia coli. The environment of the heme iron was investigated using X-ray absorption spectroscopy to determine if differences in this region were associated with the differences in reactivity. The variation in the distal side Fe-ligand distances between the two enzymes was the same within experimental error indicating that it was not the heme iron environment that produced the differences in reactivity. Analysis of variants of the E. coli catalase-peroxidase containing changes in active site residues Arg102 and His106 revealed small differences in Fe-water ligand distance including a shorter distance for the His106Tyr variant. The Arg102Leu variant was 5-coordinate, but His106Cys and Arg102Cys variants showed no changes within experimental error. These results are compared with those reported for other peroxidases.


Subject(s)
Bacterial Proteins , Catalase/chemistry , Escherichia coli/enzymology , Mycobacterium tuberculosis/enzymology , Peroxidases/chemistry , Binding Sites , Catalase/genetics , Electron Probe Microanalysis , Escherichia coli/genetics , Fourier Analysis , Heme/chemistry , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Mycobacterium tuberculosis/genetics , Peroxidases/genetics
7.
Biochemistry ; 39(19): 5868-75, 2000 May 16.
Article in English | MEDLINE | ID: mdl-10801338

ABSTRACT

Catalase-peroxidases have a predominant catalatic activity but differ from monofunctional catalases in exhibiting a substantial peroxidatic reaction which has been implicated in the activation of the antitubercular drug isoniazid in Mycobacterium tuberculosis. Hydroperoxidase I of Escherichia coli encoded by katG is a catalase-peroxidase, and residues in its putative active site have been the target of a site directed-mutagenesis study. Variants of residues R102 and H106, on the distal side of the heme, and H267, the proximal side ligand, were constructed, all of which substantially reduced the catalatic activity and, to a lesser extent, the peroxidatic activity. In addition, the heme content of the variants was reduced relative to the wild-type enzyme. The relative ease of heme loss from HPI and a mixture of tetrameric enzymes with 2, 3, and 4 hemes was revealed by mass spectrometry analysis. Conversion of W105 to either an aromatic (F) or aliphatic (I) residue caused a 4-5-fold increase in peroxidatic activity, coupled with a >99% inhibition of catalatic activity. The peroxidatic-to-catalatic ratio of the W105F variant was increased 2800-fold such that compound I could be identified by both electronic and EPR spectroscopy as being similar to the porphyrin cation radical formed in other catalases and peroxidases. Compound I, when generated by a single addition of H(2)O(2), decayed back to the native or resting state within 1 min. When H(2)O(2) was generated enzymatically in situ at low levels, active compound I was evident for up to 2 h. However, such prolonged treatment resulted in conversion of compound I to a reversibly inactivated and, eventually, to an irreversibly inactivated species, both of which were spectrally similar to compound I.


Subject(s)
Catalase/chemistry , Catalase/genetics , Escherichia coli Proteins , Escherichia coli/enzymology , Mutagenesis, Site-Directed , Peroxidases/chemistry , Peroxidases/genetics , Amino Acid Substitution/genetics , Bacterial Proteins , Binding Sites/genetics , Catalase/antagonists & inhibitors , Catalysis , Electron Spin Resonance Spectroscopy , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , Escherichia coli/genetics , Heme/chemistry , Leucine/genetics , Mass Spectrometry , Peroxidases/antagonists & inhibitors , Phenylalanine/genetics , Recombinant Proteins/chemistry , Substrate Specificity/genetics , Tryptophan/genetics
8.
J Biol Chem ; 274(39): 27717-25, 1999 Sep 24.
Article in English | MEDLINE | ID: mdl-10488114

ABSTRACT

The three-dimensional structures of two HPII variants, V169C and H392Q, have been determined at resolutions of 1.8 and 2.1 A, respectively. The V169C variant contains a new type of covalent bond between the sulfur atom of Cys(169) and a carbon atom on the imidazole ring of the essential His(128). This variant enzyme has only residual catalytic activity and contains heme b. The chain of water molecules visible in the main channel may reflect the organization of the hydrogen peroxide substrates in the active enzyme. Two alternative mechanisms, involving either compound I or free radical intermediates, are presented to explain the formation of the Cys-His covalent bond. The H392Q and H392E variants exhibit 75 and 25% of native catalytic activity, respectively. The Gln(392) variant contains only heme b, whereas the Glu(392) variant contains a mixture of heme b and cis and trans isomers of heme d, suggesting of a role for this residue in heme conversion. Replacement of either Gln(419) and Ser(414), both of which interact with the heme, affected the cis:trans ratio of spirolactone heme d. Implications for the heme oxidation mechanism and the His-Tyr bond formation in HPII are considered.


Subject(s)
Catalase/chemistry , Catalase/metabolism , Escherichia coli/enzymology , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Catalase/genetics , Crystallography, X-Ray , Cysteine , Genetic Variation , Glutamic Acid , Heme , Histidine , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Restriction Mapping
9.
Biochemistry ; 38(13): 3895-901, 1999 Mar 30.
Article in English | MEDLINE | ID: mdl-10194300

ABSTRACT

Catalase HPII from Escherichia coli is a homotetramer of 753 residue subunits. The multimer displays a number of unusual structural features, including interwoven subunits and a covalent bond between Tyr415 and His392, that would contribute to its rigidity and stability. As the temperature of a solution of HPII in 50 mM potassium phosphate buffer (pH 7) is raised from 50 to 92 degrees C, the enzyme begins to lose activity at 78 degrees C and 50% inactivation has occurred at 83 degrees C. The inactivation is accompanied by absorbance changes at 280 and 407 nm and by changes in the CD spectrum consistent with small changes in secondary structure. The subunits in the dimer structure remain associated at 95 degrees C and show a significant level of dissociation only at 100 degrees C. The exceptional stability of the dimer association is consistent with the interwoven nature of the subunits and provides an explanation for the resistance to inactivation of the enzyme. For comparison, catalase-peroxidase HPI of E. coli and bovine liver catalase are 50% inactivated at 53 and 56 degrees C, respectively. In 5.6 M urea, HPII exhibits a coincidence of inactivation, CD spectral change, and dissociation of the dimer structure with a midpoint of 65 degrees C. The inactive mutant variants of HPII which fold poorly during synthesis and which lack the Tyr-His covalent bond undergo spectral changes in the 78 to 84 degrees C range, revealing that the extra covalent linkage is not important in the enhanced resistance to denaturation and that problems in the folding pathway do not affect the ultimate stability of the folded structure.


Subject(s)
Catalase/chemistry , Catalase/metabolism , Escherichia coli/enzymology , Catalase/genetics , Circular Dichroism , Enzyme Activation/genetics , Enzyme Stability/genetics , Guanidine , Hot Temperature , Models, Molecular , Mutagenesis, Site-Directed , Protein Denaturation , Sodium Dodecyl Sulfate , Urea
10.
Protein Sci ; 8(3): 490-8, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10091651

ABSTRACT

The heme-containing catalase HPII of Escherichia coli consists of a homotetramer in which each subunit contains a core region with the highly conserved catalase tertiary structure, to which are appended N- and C-terminal extensions making it the largest known catalase. HPII does not bind NADPH, a cofactor often found in catalases. In HPII, residues 585-590 of the C-terminal extension protrude into the pocket corresponding to the NADPH binding site in the bovine liver catalase. Despite this difference, residues that define the NADPH pocket in the bovine enzyme appear to be well preserved in HPII. Only two residues that interact ionically with NADPH in the bovine enzyme (Asp212 and His304) differ in HPII (Glu270 and Glu362), but their mutation to the bovine sequence did not promote nucleotide binding. The active-site heme groups are deeply buried inside the molecular structure requiring the movement of substrate and products through long channels. One potential channel is about 30 A in length, approaches the heme active site laterally, and is structurally related to the branched channel associated with the NADPH binding pocket in catalases that bind the dinucleotide. In HPII, the upper branch of this channel is interrupted by the presence of Arg260 ionically bound to Glu270. When Arg260 is replaced by alanine, there is a threefold increase in the catalytic activity of the enzyme. Inhibitors of HPII, including azide, cyanide, various sulfhydryl reagents, and alkylhydroxylamine derivatives, are effective at lower concentration on the Ala260 mutant enzyme compared to the wild-type enzyme. The crystal structure of the Ala260 mutant variant of HPII, determined at 2.3 A resolution, revealed a number of local structural changes resulting in the opening of a second branch in the lateral channel, which appears to be used by inhibitors for access to the active site, either as an inlet channel for substrate or an exhaust channel for reaction products.


Subject(s)
Catalase/metabolism , Escherichia coli/enzymology , Alanine/genetics , Base Sequence , Binding Sites , Biopolymers , Catalase/antagonists & inhibitors , Catalase/chemistry , Catalase/genetics , DNA Primers , Enzyme Inhibitors/pharmacology , Mutagenesis, Site-Directed , NADP/metabolism , Protein Conformation
11.
FEMS Microbiol Lett ; 170(2): 307-12, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-10068295

ABSTRACT

The catalase-peroxidase hydroperoxidase I of Escherichia coli has been confirmed to be located in the cytoplasm using two independent methods. Catalase activity was found predominantly (> 95%) in the cytoplasmic fraction following spheroplast formation. The cytoplasmic enzyme glucose-6-phosphate dehydrogenase and the periplasmic enzyme alkaline phosphatase were used as controls. The second method of immunogold staining for the enzyme in situ revealed an even distribution of the enzyme across the cell.


Subject(s)
Bacterial Proteins/metabolism , Catalase/metabolism , Cytoplasm/enzymology , Escherichia coli Proteins , Escherichia coli/enzymology , Escherichia coli/growth & development , Immunohistochemistry
12.
Proteins ; 34(2): 155-66, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-10022351

ABSTRACT

Catalase HPII from Escherichia coli, a homotetramer of subunits with 753 residues, is the largest known catalase. The structure of native HPII has been refined at 1.9 A resolution using X-ray synchrotron data collected from crystals flash-cooled with liquid nitrogen. The crystallographic agreement factors R and R(free) are respectively 16.6% and 21.0%. The asymmetric unit of the crystal contains a whole molecule that shows accurate 222-point group symmetry. The structure of the central part of the HPII subunit gives a root mean square deviation of 1.5 A for 477 equivalencies with beef liver catalase. Most of the additional 276 residues of HPII are located in either an extended N-terminal arm or in a C-terminal domain organized with a flavodoxin-like topology. A small number of mostly hydrophilic interactions stabilize the relative orientation between the C-terminal domain and the core of the enzyme. The heme component of HPII is a cis-hydroxychlorin gamma-spirolactone in an orientation that is flipped 180 degrees with respect to the orientation of the heme found in beef liver catalase. The proximal ligand of the heme is Tyr415 which is joined by a covalent bond between its Cbeta atom and the Ndelta atom of His392. Over 2,700 well-defined solvent molecules have been identified filling a complex network of cavities and channels formed inside the molecule. Two channels lead close to the distal side heme pocket of each subunit suggesting separate inlet and exhaust functions. The longest channel, that begins in an adjacent subunit, is over 50 A in length, and the second channel is about 30 A in length. A third channel reaching the heme proximal side may provide access for the substrate needed to catalyze the heme modification and His-Tyr bond formation. HPII does not bind NADPH and the equivalent region to the NADPH binding pocket of bovine catalase, partially occluded in HPII by residues 585-590, corresponds to the entrance to the second channel. The heme distal pocket contains two solvent molecules, and the one closer to the iron atom appears to exhibit high mobility or low occupancy compatible with weak coordination.


Subject(s)
Bacterial Proteins/chemistry , Catalase/chemistry , Escherichia coli/enzymology , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Heme/chemistry , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , NADP/chemistry , Protein Conformation , Water/chemistry
13.
Can J Microbiol ; 44(8): 707-17, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9830102

ABSTRACT

In Escherichia coli, the transcription factor sigma s, encoded by rpoS, controls the expression of a large number of genes involved in cellular responses to a diverse number of stresses, including starvation, osmotic stress, acid shock, cold shock, heat shock, oxidative DNA damage, and transition to stationary phase. A list of over 50 genes under the control of rpoS has been compiled. The transcription factor sigma s acts predominantly as a positive effector, but it does have a negative effect on some genes. The synthesis and accumulation of sigma s are controlled by mechanisms affecting transcription, translation, proteolysis, and the formation of the holoenzyme complex. Transcriptional control of rpoS involves guanosine 3',5'-bispyrophosphate (ppGpp) and polyphosphate as positive regulators and the cAMP receptor protein-cAMP complex (CRP-cAMP) as a negative regulator. Translation of rpoS mRNA is controlled by a cascade of interacting factors, including Hfq, H-NS, dsrA RNA, LeuO, and oxyS RNA that seem to modulate the stability of a region of secondary structure in the ribosome-binding region of the gene's mRNA. The transcription factor sigma s is sensitive to proteolysis by ClpPX in a reaction that is promoted by RssB and inhibited by the chaperone DnaK. Despite the demonstrated involvement of so many factors, arguments have been presented suggesting that sensitivity to proteolysis may be the single most important modulator of sigma s levels. The activity of sigma s may also be modulated by trehalose and glutamate, which activate holoenzyme formation and promote holoenzyme binding to certain promoters.


Subject(s)
Bacterial Proteins/genetics , Escherichia coli/genetics , Regulon , Sigma Factor/genetics , Bacterial Proteins/metabolism , Base Sequence , DNA, Bacterial/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Genes, Bacterial , Holoenzymes/biosynthesis , Promoter Regions, Genetic , Protein Biosynthesis , Protein Processing, Post-Translational , Sigma Factor/metabolism
14.
Protein Eng ; 11(7): 549-55, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9740372

ABSTRACT

The subunit of catalase HPII from Escherichia coli is 753 residues in length and contains a core of approximately 500 residues, with high structural similarity to all other heme catalases. To this core are added extensions of approximately 80 and 180 residues at the N- and C-termini, respectively. The tetrameric structure is made up of a pair of interwoven dimers in which 90 N-terminal residues of each subunit are inserted through a loop formed by the hinge region linking the beta-barrel and alpha-helical domains of the adjacent subunit. A high concentration of proline residues is found in the vicinity of the overlap regions. To study the influence of the extended regions on folding and subunit association of HPII, a diversity of modifications have been introduced. Removal of the complete C-terminal domain or the N-terminal extension, either separately or together, effectively creating a small subunit catalase, resulted in no enzyme accumulation. Systematic truncations showed that only nine C-terminal residues (Ile745 to Ala753) could be removed without significantly affecting the accumulation of active enzyme. Removal or even conservative replacements of the side chain of Arg744 significantly reduced the accumulation of active enzyme despite this residue interacting only with the C-terminal domain. Removal of as few as 18 residues from the N-terminus also reduced accumulation of active enzyme. Changes to other residues in the protein, including residues in the heme binding pocket, also reduced the accumulation of active protein without substantially affecting the enzyme specific activity. Implications of these data for the interdependence of subunit folding and subunit-subunit interactions are discussed.


Subject(s)
Catalase/genetics , Escherichia coli/genetics , Mutation , Catalase/biosynthesis , Protein Folding , Structure-Activity Relationship
15.
Mol Biol Evol ; 14(9): 951-8, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9287428

ABSTRACT

Seventy-four catalase protein sequences, including 29 bacterial, 8 fungal, 7 animal, and 30 plant sequences, were compiled, and 70 were used for phylogenetic reconstruction. The core of the resulting tree revealed unique, separate groups of plant and animal catalases, two groups of fungal catalases, and three groups of bacterial catalases. The only overlap of kingdoms occurred within one branch and involved fungal and bacterial large-subunit enzymes. The other fungal branch was closely linked to the group of animal enzymes. Group I bacterial catalases were more closely related to the plant enzymes and contained such diverse taxa as the Gram-positive Listeria seeligeri, Deinocococcus radiodurans, and gamma-proteobacteria. Group III bacterial sequences were more closely related to fungal and animal sequences and included enzymes from a broad range of bacteria including high- and low-GC Gram positives, proteobacteria, and a bacteroides species. Group II was composed of large-subunit catalases from diverse sources including Gram positives (low-GC Bacilli and high-GC Mycobacteria), proteobacteria, and species of the filamentous fungus Aspergillus. These data can be interpreted in terms of two gene duplication events that produced a minimum of three catalase gene family members that subsequently evolved in response to environmental demands. Horizontal gene transfer may have been responsible for the group II mixture of bacterial and fungal large-subunit catalases.


Subject(s)
Catalase/genetics , Phylogeny , Amino Acid Sequence , Animals , Bacteria/enzymology , Base Composition , DNA/chemistry , Fungi/enzymology , Molecular Sequence Data , Mutation/genetics , Plants/enzymology
16.
Biochim Biophys Acta ; 1352(2): 161-6, 1997 May 30.
Article in English | MEDLINE | ID: mdl-9199247

ABSTRACT

The rpoS gene of Escherichia coli encodes an alternative sigma factor of RNA polymerase sigma38 (or sigma(s)) that is required for transcription of katE encoding catalase HPII. The transcription start site of the single katE transcript identified by ribonuclease protection has been determined by primer extension analysis to be either 53 or 54 bp (depending on the strain used) upstream of the open reading frame. A series of promoter fragments were constructed and fused to lacZ to confirm the start site location. A - 10 sequence similar to that found in other sigma70- and sigma38-dependent E. coli promoters was identified 8 or 7 bp upstream of the start site but a sigma70-dependent -35 sequence was not evident.


Subject(s)
Bacterial Proteins/genetics , Catalase/genetics , Escherichia coli/genetics , Sigma Factor/genetics , Amino Acid Sequence , Base Sequence , Molecular Sequence Data , Polymerase Chain Reaction , Promoter Regions, Genetic
17.
Protein Sci ; 6(5): 1016-23, 1997 May.
Article in English | MEDLINE | ID: mdl-9144772

ABSTRACT

A bond between the N delta of the imidazole ring of His 392 and the C beta of the essential Tyr 415 has been found in the refined crystal structure at 1.9 A resolution of catalase HPII of Escherichia coli. This novel type of covalent linkage is clearly defined in the electron density map of HPII and is confirmed by matrix-assisted laser desorption/ionization mass spectrometry analysis of tryptic digest mixtures. The geometry of the bond is compatible with both the sp3 hybridization of the C beta atom and the planarity of the imidazole ring. Two mutated variants of HPII active site residues, H128N and N201H, do not contain the His 392-Tyr 415 bond, and their crystal structures show that the imidazole ring of His 392 was rotated, in both cases, by 80 degrees relative to its position in HPII. These mutant forms of HPII are catalytically inactive and do not convert heme b to heme d, suggesting a relationship between the self-catalyzed heme conversion reaction and the formation of the His-Tyr linkage. A model coupling the two processes and involving the reaction of one molecule of H2O2 on the proximal side of the heme with compound 1 is proposed.


Subject(s)
Catalase/chemistry , Escherichia coli/enzymology , Histidine , Protein Conformation , Tyrosine , Amino Acid Sequence , Binding Sites , Catalase/isolation & purification , Catalase/metabolism , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thermodynamics , Trypsin
18.
Biochim Biophys Acta ; 1298(2): 241-9, 1996 Dec 05.
Article in English | MEDLINE | ID: mdl-8980649

ABSTRACT

Cyanide forms an inhibitory complex with the haem d-containing E. coli catalase HPII, spectrally similar to the cyanide complex of beef liver enzyme but with absorption bands shifted 90 nm towards the red end of the spectrum. Both the Kd and Ki values are approximately 7 microM in the wild-type enzyme. The cyanide reaction is slow, with a bimolecular 'on' constant approx. 2000 x smaller than that of eukaryotic enzyme, and an 'off' constant diminished by a similar amount. Catalases with a mutated distal histidine (H128) fail to bind cyanide at cyanide concentrations below 50 mM. Catalases with a mutated distal asparagine (N201) show only small changes in cyanide affinity from the wild type. The major fraction of HPII N201A has a Kd approximately 40 microM, and a minor fraction has a lower cyanide affinity; the major fraction of HPII N201Q has a Kd approximately 15 microM. The Kd and Ki for HPII N201D is approximately 8 microM, essentially identical with that of the wild type but N201D appears to bind cyanide somewhat more rapidly than does wild-type enzyme. The HPII mutant N201H can be obtained in both haem d and protohaem forms; it exhibits two types of cyanide binding behaviour. In its protohaem form it binds cyanide poorly (Kd > or = 0.25 mM). After peroxide treatment converts t into haem d or a closely related species it binds cyanide with a much higher affinity (Kd approximately 15 microM). Cyanide binding to HPII requires a distal histidine to provide hydrogen-bonding stability, but not a distal asparagine. Rates of cyanide binding and release are controlled by haem group accessibility through the channel leading to the outside. In HPII N201H channel opening may depend upon oxidation of the haem from the starting protohaem to the final haem d form.


Subject(s)
Catalase/metabolism , Cyanides/metabolism , Escherichia coli/enzymology , Animals , Catalase/antagonists & inhibitors , Catalase/genetics , Cattle , Kinetics , Ligands , Liver/enzymology , Mutagenesis, Site-Directed , Spectrum Analysis
19.
J Biol Chem ; 271(15): 8863-8, 1996 Apr 12.
Article in English | MEDLINE | ID: mdl-8621527

ABSTRACT

A heme d prosthetic group with the configuration of a cis-hydroxychlorin gamma-spirolactone has been found in the crystal structures of Penicillium vitale catalase and Escherichia coli catalase hydroperoxidase II (HPII). The absolute stereochemistry of the two heme d chiral carbon atoms has been shown to be identical. For both catalases the heme d is rotated 180 degrees about the axis defined by the alpha-gamma-meso carbon atoms, with respect to the orientation found for heme b in beef liver catalase. Only six residues in the heme pocket, preserved in P. vitale and HPII, differ from those found in the bovine catalase. In the crystal structure of the inactive N201H variant of HPII catalase the prosthetic group remains as heme b, although its orientation is the same as in the wild type enzyme. These structural results confirm the observation that heme d is formed from protoheme in the interior of the catalase molecule through a self-catalyzed reaction.


Subject(s)
Catalase/chemistry , Heme/chemistry , Crystallography, X-Ray , Escherichia coli/enzymology , Hemeproteins/chemistry , Hydrogen Bonding , Models, Molecular , Molecular Structure , Penicillium/enzymology
20.
Arch Biochem Biophys ; 323(2): 438-46, 1995 Nov 10.
Article in English | MEDLINE | ID: mdl-7487109

ABSTRACT

The physical properties and activities of the purified catalase-peroxidase hydroperoxidase I (HPI) of Escherichia coli (EcHPI) and HPI with a carboxyl-terminal extension of Mycobacterium tuberculosis (MtHPI-e) are compared to those of commercial preparations of horseradish peroxidase (HRP). The catalase-peroxidase proteins had similar absorption spectra and differed primarily in that MtHPI-e has a higher peroxidatic to catalatic activity ratio than EcHPI. Trypsin cleavage of MtHPI-e resulted in the formation of an active catalase-peroxidase lacking the carboxyl-terminal extension. The three enzymes, HRP, MtHPI-e, and EcHPI, mediated the isoniazid- and H2O2-dependent production of radical species, as detected by nitroblue tetrazolium reduction. A constant flux of H2O2, generated in situ from glucose oxidase and glucose was used. MtHPI-e was more effective at isoniazid-dependent radical production than EcHPI and HRP. Similar qualitative results were obtained by staining nondenaturing polyacrylamide gels for activity with nitroblue tetrazolium in the presence of isoniazid and H2O2. The absorbance spectrum of HRP exhibited changes during incubation with isoniazid and H2O2 consistent with the formation of several typical reaction intermediates, whereas the catalase-peroxidases exhibited no distinct spectral changes. The results suggest that the sensitivity of M. tuberculosis to isoniazid may be the result of isoniazid-dependent radical formation by the catalase-peroxidase in the absence of other catalase activities to remove substrate H2O2.


Subject(s)
Escherichia coli Proteins , Isoniazid/metabolism , Peroxidases/metabolism , Bacterial Proteins , Catalase/metabolism , Escherichia coli/enzymology , Free Radicals , Horseradish Peroxidase/metabolism , Hydrogen Peroxide/metabolism , Kinetics , Mycobacterium tuberculosis/enzymology , Spectrum Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...